ArticlePDF Available

Cardiovascular events in patients with COPD: TORCH study results

Authors:
  • Pulmonary Research Institute of Southeast Michigan

Abstract and Figures

Previous studies have suggested that long-term use of beta agonists to treat chronic obstructive pulmonary disease (COPD) may increase the risk of cardiovascular adverse events. In this post hoc analysis, data from the TOwards a Revolution in COPD Health (TORCH) study were used to investigate whether use of the long-acting beta(2) agonist salmeterol over 3 years increased the risk of cardiovascular adverse events in patients with moderate to severe COPD. TORCH was a randomised, double-blind, placebo controlled study conducted at 444 centres in 42 countries. Patients (n=6184; safety population) received twice daily combined salmeterol 50 microg plus fluticasone propionate 500 microg (SFC), either component alone, or placebo. Adverse events were recorded every 12 weeks for 3 years. The probability of having a cardiovascular adverse event by 3 years was 24.2% for placebo, 22.7% for salmeterol, 24.3% for fluticasone propionate and 20.8% for SFC. Although a history of myocardial infarction doubled the probability of cardiovascular adverse events, the event rates remained similar across treatment groups. Post hoc analysis of the 3-year TORCH dataset showed that salmeterol alone or in combination (SFC) did not increase the risk of cardiovascular events in patients with moderate to severe COPD.
Content may be subject to copyright.
Cardiovascular events in patients with COPD: TORCH
Study results
Peter M A Calverley,
1
Julie A Anderson,
2
Bartolome Celli,
3
Gary T Ferguson,
4
Christine Jenkins,
5
Paul W Jones,
6
Courtney Crim,
7
Lisa R Willits,
2
Julie C Yates,
7
Jørgen Vestbo,
8
on behalf of the TORCH Investigators
ABSTRACT
Background Previous studies have suggested that long-
term use of
b
agonists to treat chronic obstructive
pulmonary disease (COPD) may increase the risk of
cardiovascular adverse events. In this post hoc analysis,
data from the TOwards a Revolution in COPD Health
(TORCH) study were used to investigate whether use of
the long-acting
b
2
agonist salmeterol over 3 years
increased the risk of cardiovascular adverse events in
patients with moderate to severe COPD.
Methods TORCH was a randomised, double-blind,
placebo controlled study conducted at 444 centres in 42
countries. Patients (n¼6184; safety population) received
twice daily combined salmeterol 50
m
g plus fluticasone
propionate 500
m
g (SFC), either component alone, or
placebo. Adverse events were recorded every 12 weeks
for 3 years.
Results The probability of having a cardiovascular
adverse event by 3 years was 24.2% for placebo, 22.7%
for salmeterol, 24.3% for fluticasone propionate and
20.8% for SFC. Although a history of myocardial
infarction doubled the probability of cardiovascular
adverse events, the event rates remained similar across
treatment groups.
Conclusion Post hoc analysis of the 3-year TORCH
dataset showed that salmeterol alone or in combination
(SFC) did not increase the risk of cardiovascular events in
patients with moderate to severe COPD.
Chronic obstructive pulmonary disease (COPD) is
now recognised as being a highly prevalent condi-
tion
1
that causes signicant morbidity and
mortality,
2
and commonly coexists with cardiovas-
cular disease.
3
Cardiovascular events are one of the
leading causes of mortality and hospitalisation in
patients with COPD, particularly in those with mild
to moderate disease.
45
Moreover, COPD is a strong
risk factor for cardiovascular events, independent of
smoking,
367
and impaired lung function is an
important marker for cardiovascular mortality.
89
However, it is unclear whether COPD interventions
other than smoking cessation
4
can modify the
increased cardiovascular risk associated with COPD.
Recently there has been concern that the long-
term use of inhaled bronchodilators commonly
used in the treatment of COPD, including long-
acting
b
2
agonist (LABA) and anticholinergic drugs,
may increase the risk of cardiovascular
complications.
10e14
However, prospective data
about the relative risk of therapy in patients with
sufcient symptoms to be offered treatment with
these drugs has, until recently, been lacking.
While the 3-year TOwards a Revolution in
COPD Health (TORCH) study primarily
investigated the effect of combination therapy with
the LABA salmeterol (SAL) and the inhaled corti-
costeroid uticasone propionate (FP) compared
with placebo on all-cause mortality, other efcacy
outcomes and adverse events (AEs) were also
measured. The primary paper including the
mortality analysis has already been published.
15
It
reported that long-term use of SAL or the SAL plus
FP combination (SFC) did not increase the rate of
cardiac death. In view of the continuing concerns
about cardiovascular safety in COPD therapy, we
extended this analysis (post hoc) to consider the
occurrence of cardiovascular AEs and serious
adverse events (SAEs) in the TORCH study. We
also explored the factors that might determine the
incidence of cardiovascular events in these patients.
Some of these results have previously been
presented in abstract form.
16
METHODS
Patients
Details of patient eligibility and study entry criteria
have been published previously.
15
Eligible patients
were current or former smokers aged 40e80 years
with a prebronchodilator forced expiratory volume
in 1 s (FEV
1
)<60% of the predicted value and
a ratio of prebronchodilator FEV
1
to forced vital
capacity (FVC) of #0.70. The only exclusion
criterion with respect to comorbidities was that
subjects were considered unlikely to die of some-
thing other than COPD in the subsequent 3 years.
Design overview
The study design has been described in detail
previously.
15 17
TORCH was a multicentre, rand-
omised, double-blind, parallel-group, placebo
controlled study conducted at 444 centres in 42
countries. After a 2-week run-in period, eligible
patients were stratied by smoking status and
randomised to receive either SFC 50/500
m
g, SAL
50
m
g, FP 500
m
g or placebo twice daily for 3 years
via a Diskus/Accuhaler inhaler (GlaxoSmithKline,
Greenford, UK). Full details of the randomisation
procedure have been reported elsewhere.
15 17
The primary efcacy end point of the TORCH
study was all-cause mortality. Other end points
were rate of COPD exacerbations, health-related
quality of life, lung function and AEs. After
randomisation, patient visits occurred every
12 weeks to record any AE. At each visit the subject
was allowed to spontaneously mention any
<Supplementary tables are
published online only. To view
these files please visit the
journal online (http://thorax.bmj.
com).
1
University Hospital Aintree,
Liverpool, UK
2
GlaxoSmithKline, Middlesex,
UK
3
Caritas-St Elizabeth’s Medical
Center, Brighton,
Massachusetts, USA
4
Pulmonary Research Institute
of Southeast Michigan, Livonia,
Michigan, USA
5
Woolcock Institute of Medical
Research, NSW, Australia
6
University of London, London,
UK
7
GlaxoSmithKline, Research
Triangle Park, North Carolina,
USA
8
Hvidovre Hospital, Hvidovre,
Denmark and Wythenshawe
Hospital, Manchester, UK
Correspondence to
Professor P M A Calverley,
Department of Medicine,
University Hospital Aintree,
Longmoor Lane, Liverpool
L9 7AL, UK;
pmacal@liverpool.ac.uk
Received 28 January 2010
Accepted 19 April 2010
Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077 719
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
problems, then the investigator asked the following standard
questions: (1) Have you had any (other) medical problems since
your last visit/assessment?and (2) Have you taken any new
medicines other than those given to you within this study since
your last visit/assessment?An AE was dened as any unfav-
ourable and unintended sign (including a clinically signicant
abnormal laboratory nding), symptom or disease temporally
associated with the use of the blinded study product, whether or
not it was considered to be related to that product.
SAEs were those that resulted in any of the following
outcomes: (1) death; (2) an immediate risk of death; (3) hospi-
talisation or prolongation of an existing hospitalisation; or (4)
any other important medical event which, in the opinion of the
investigator, jeopardised the subjects health. An independent
safety and efcacy data monitoring committee performed safety
reviews of all SAEs every 6 months. Causes of death were
independently adjudicated in a standardised fashion by a clinical
end point committee.
18
Cardiovascular safety evaluation
Cardiovascular AEs were classied based on the Medical
Dictionary for Regulatory Activities (MedDRA) Version 8.1
terms. MedDRA is the AE classication dictionary endorsed by
the International Conference on Harmonisation of Technical
Requirements for Registration of Pharmaceuticals for Human
Use (ICH). All events in the cardiac disorders system organ class
plus selected events from the vascular disorders and nervous
system disorders system organ classes were included (table 1).
Further analysis was also conducted on the subset of ischaemic
events and on events related to stroke (table 1). No specic
information was collected about whether patient-reported AEs
had been objectively veried.
Subjects receiving medications commonly used to treat
cardiovascular disease were identied; these medications were
selected from dictionary groupings (listed in table 1).
Statistical analysis
The study was powered for its primary all-cause mortality end
point and was not formally powered to detect differences in
numbers of AEs between treatments.
The safety population included all patients who took at least
one dose of study medication. The number and proportion of
patients reporting a cardiovascular AE over the 3 years were
summarised by treatment group. To correct for differential
treatment exposure between the treatments, the rate of cardiac
events per 1000 treatment years was calculated by dividing the
total number of AEs by the total number of years patients were
exposed to study treatment, then multiplying by 1000. On-
treatment deaths were dened as any death occurring up to
14 days after patients stopped their study medication.
The time to rst cardiovascular AE was compared between
treatment groups using KaplaneMeier estimates and the log-
rank test, stratied by smoking status; KaplaneMeier cumula-
tive incidence curves were also generated. Statistical signicance
was set at p<0.05.
RESULTS
Study population
Of 8554 patients with COPD recruited, 6184 were randomised
and evaluated for safety (gure 1). This included 72 patients
from ve sites excluded from the published efcacy analyses.
15
One subject was randomised to placebo but took FP for the
majority of the treatment period and was analysed with the FP
group.
Demographic and baseline patient characteristics were
balanced across treatment groups (table 2). The mean age was
65 years, 76% were male, mean smoking history was 48 pack-
years and baseline postbronchodilator FEV
1
was 44% of
predicted. At baseline, 7% of patients reported a history of
previous myocardial infarction (MI), 41% were taking cardio-
vascular medications and 60% were taking short-acting anti-
cholinergic drugs. Use of the long-acting anticholinergic
tiotropium bromide during the study was low (3%) and was
similar across treatment groups. Of the 187 subjects receiving
tiotropium, over half took it for 12 weeks or less.
The proportion of patients who withdrew from the study was
highest in the placebo group (44%) and lowest in the SFC group
(34%) (SAL 37%, FP 39%). The total number of patient-years of
exposure to the study drugs was 3278 for placebo, 3531 for SAL,
3555 for FP and 3700 for SFC.
Cardiovascular AEs
The proportion of patients who experienced a cardiovascular AE
or cardiovascular SAE during the study was similar across
treatment groups (17e20% and 10e12%, respectively) (table 3).
When expressed as the number of events per 1000 treatment
years, the rate of cardiovascular AEs was 142 in the placebo
group and 110 in the SFC group. The probability of patients
having a cardiovascular AE by 3 years was lowest for SFC
at 20.8% (24.2% for placebo, 22.7% for SAL and 24.3% for FP;
gure 2A; table 3). The probability of patients having a cardio-
vascular SAE by 3 years was lowest for SFC at 12.5% (15.4% for
placebo, 13.6% for SAL and 14.7% for FP; table 3).
The proportion of patients experiencing an ischaemic cardio-
vascular AE was similar across treatment groups (9e11%;
table 4). The rate of ischaemic cardiovascular AEs per 1000
treatment years was 68 for placebo, 70 for SAL, 62 for FP and 54
for SFC. The probability of patients having an ischaemic cardio-
vascular AE by 3 years was lowest for SFC at 11.3%, 14.6% for
placebo, 13.4% for SAL and 13.8% for FP (gure 2B and table 4).
The proportion of patients with a stroke-related AE over
the 3-year course of the study was similar in each treatment
group (3% placebo, 2% SAL, 3% FP and 3% SFC). The rate of
Table 1 List of MedDRA system organ class (SOC) high-level group
terms (HLGT) used for analysis of cardiovascular events, ischaemic
cardiovascular events and stroke events, and list of cardiovascular
medications included in the analysis
Cardiovascular events
<Cardiac disorders (SOC)
– Coronary artery disorders
– Cardiac arrhythmias
– Heart failures
– Cardiac disorder signs and symptoms
– Myocardial disorders
– Cardiac valve disorders
– Pericardial disorders
<Nervous system disorders (SOC)
– Central nervous system vascular disorders
<Vascular disorders (SOC)
– Arteriosclerosis, stenosis, vascular
insufficiency and necrosis
– Aneurysms and artery dissections
– Embolism and thrombosis
Ischaemic events
<Ischaemic coronary artery
disorders
<Coronary artery disorders NEC
<Heart failures NEC
<Right ventricular failures
<Left ventricular failures
<Cardiomyopathies
<Pericardial disorders NEC
<Non-infectious pericarditis
Stroke events
<Central nervous system
vascular disorders (HLGT)
Cardiovascular medications
<ACE inhibitors
<Angiotensin II antagonists
<Antihypertensives
<Beta blockers
<Calcium channel blockers
<Antiarrythmics
<Cardiac glycosides
<Adrenergic and dopaminergic
agents
<Organic nitrates
NEC, not elsewhere classified.
720 Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
stroke-related AEs per 1000 treatment years was 17 for placebo,
13 for SAL, 16 for FP and 12 for SFC.
The likelihood of experiencing a cardiovascular AE was unaf-
fected by gender and current smoking status (table 5). Patients
who were on cardiovascular medications at baseline (gure 3),
reported a previous MI (gure 4), were older or had lower
baseline FEV
1
(table 5) had a higher probability of having
a cardiovascular AE. There were no signicant differences
between treatments in the likelihood of an event being reported,
nor was there consistent evidence for an interaction between
treatment and spirometrically-dened disease severity with
respect to cardiac events (see table 1 in online supplement) . In
patients who had previously had an MI, the likelihood of having
a cardiovascular event over 3 years was 54.4% on salmeterol
compared with 49.1% with placebo (p ¼0.51) and 44.1% with
SFC (p¼0.051 relative to salmeterol (gure 4B and table 5).
Figure 1 Patient flow. *The number of
patients who underwent randomisation
and the number of those included in the
safety population differ in the placebo
group and the fluticasone propionate
group because one patient who was
assigned to placebo received
fluticasone propionate for more than
half the study period; this patient was
therefore included in the fluticasone
propionate group for the safety
analysis. AE, adverse event.
Table 2 Patient demographic and baseline characteristics of the safety population
Variable
Placebo
(n[1544)
Salmeterol
(n [1542)
FP
(n [1552)
SFC
(n [1546)
Age at enrolment, years 65.1 (8.1) 65.2 (8.2) 65.1 (8.4) 65.0 (8.3)
Male gender, n (%) 1175 (76) 1176 (76) 1169 (75) 1164 (75)
Body mass index, kg/m
2
25.5 (5.2) 25.4 (5.2) 25.3 (5.1) 25.4 (5.3)
Postbronchodilator FEV
1
, % predicted 44.1 (12.2) 43.6 (12.6) 44.1 (12.3) 44.3 (12.4)
<30%, n (%) 215 (14) 261 (17) 223 (14) 246 (16)
30e<50%, n (%) 786 (51) 750 (49) 785 (51) 735 (48)
$50%, n (%) 543 (35) 531 (34) 544 (35) 565 (37)
Geographical region, n (%)
USA 348 (23) 351 (23) 350 (23) 352 (23)
Asia Pacific 188 (12) 189 (12) 193 (12) 188 (12)
Eastern Europe 297 (19) 296 (19) 293 (19) 293 (19)
Western Europe 478 (31) 475 (31) 481 (31) 477 (31)
Other 234 (15) 231 (15) 234 (15) 236 (15)
Current smoker, n (%) 664 (43) 657 (43) 666 (43) 667 (43)
Pack-years smoked 48.6 (27.0) 49.3 (27.7) 49.2 (28.5) 46.9 (26.5)
Previous COPD treatment, n (%)*
ICS alone 346 (22) 278 (18) 310 (20) 295 (19)
LABA alone 118 (8) 138 (9) 133 (9) 137 (9)
ICS + LABA 453 (29) 417 (27) 416 (27) 437 (28)
Neither ICS nor LABA 557 (36) 634 (41) 629 (41) 623 (40)
Previous MI, n (%) n¼1543 n¼1541 n¼1552 n¼1545
0 1432 (93) 1427 (93) 1460 (94) 1443 (93)
1 91 (6) 93 (6) 79 (5) 83 (5)
$2 29 (1) 21 (1) 13 (<1) 19 (1)
Baseline CV treatment, n (%)y608 (39%) 630 (41%) 630 (41%) 662 (43%)
Baseline short-acting anticholinergics, n (%) 926 (60%) 921 (60%) 943 (61%) 925 (60%)
Data are mean (SD) unless otherwise indicated.
CV, cardiovascular; FEV
1
, forced expiratory volume in 1 s; FP, fluticasone propionate; ICS, inhaled corticosteroid; LABA, long-acting
b
2
agonist; MI, myocardial infarction; SFC, salmeterol/fluticasone propionate combination.
*Self-reported in the 12 months prior to screening.
ySee list in table 1.
Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077 721
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
However, the pattern of risk in other groups was inconsistent
with salmeterol-treated patients having a similar incidence of
events to patients treated with FP, as identied by their use of
previous cardiac therapies (gure 3B). Patients who were taking
short-acting anticholinergic treatment at baseline had a higher
probability of cardiovascular events (table 5); however, these
patients also had lower baseline percentage predicted FEV
1
. The
pattern of cardiovascular events was similar across treatment
groups irrespective of baseline anticholinergic use.
Cardiovascular deaths
There were 882 deaths (14%) during the 3-year study period,
including those both on and off study treatment (see table 2 in
online supplement). Of these, 239 were due to cardiovascular
causes as adjudicated by the clinical end point committee. For
placebo, 71 deaths (4.6%) were due to cardiovascular causes
compared with 45 (2.9%) for SAL, 61 (3.9%) for FP and 62 (4.0%)
for SFC. There were 468 deaths while on treatment, of which
172 were due to cardiovascular causes. For placebo, 47 (3.0%)
deaths were due to cardiovascular causes compared with 33
(2.1%) for SAL, 43 (2.8%) for FP and 49 (3.2%) for SFC.
DISCUSSION
COPD is not simply a lung disease
19
but is associated with an
increased likelihood of complications outside the lungs. Support
for this concept comes from recent studies that have shown
signicantly increased pulse wave velocity, independent of
smoking status and other risk factors, in patients with stable
COPD.
20
In these circumstances it is no surprise that cardio-
vascular morbidity and mortality is high in COPD.
Both
b
agonists and antimuscarinic agents, the main classes of
bronchodilator drugs used to treat COPD, have the potential to
precipitate cardiac rhythm disturbances and other cardiac
events; however, this has not been regarded as important in
clinical practice until recently. Unfortunately, unlike the situa-
tion for cardiovascular disease, most studies of drug treatment in
COPD have been relatively brief (#1 year) and have only
reported on-treatment data. These studies have made the largest
contribution to the systematic reviews in this eld, at least as far
as patients receiving treatment for symptoms are concerned. As
the TORCH study was conducted in a patient group likely to be
prescribed inhaled LABAs, the TORCH dataset addresses some
of the problems inherent in these earlier analyses. The current
analysis provides generally reassuring results about the cardio-
vascular safety of inhaled LABA treatments in patients with
COPD.
TORCH is the largest and longest prospective trial to examine
the role of an inhaled LABA and an inhaled corticosteroid in
COPD. Half of the >6000 patients were randomised to a regime
containing SAL and, allowing for dropouts, this provides 7231
patient-years of exposure to these agents. Over the 3 years,
approximately 1 in 5 patients experienced a cardiovascular AE.
The event rate was lowest in those receiving SAL in combination
with FP and not different from those patients treated with
placebo or with LABA monotherapy. A SAE requiring hospital-
isation and new cardiovascular ischaemic events were approxi-
mately half as common as the total cardiovascular event rate,
Figure 2 Cumulative incidence of (A) all cardiovascular adverse events
and (B) ischaemic cardiovascular adverse events. Full details of the data
presented in (A) and (B) including statistical testing are shown in tables
3 and 4, respectively. Vertical bars represent standard errors. AE,
adverse event; FP, fluticasone propionate; SAL, salmeterol; SFC,
salmeterol/fluticasone propionate combination.
Table 3 Summary of all cardiovascular AEs and SAEs
Placebo
(n[1544)
Salmeterol
(n[1542)
FP
(n[1552)
SFC
(n[1546)
All CV AEs
Patients with CV AE, n (%) 281 (18) 287 (19) 306 (20) 267 (17)
Rate* of CV AE (no of events) 142 (466) 141 (496) 130 (462) 110 (405)
Probabilityyof CV AE by
3 years, %
24.2 22.7 24.3 20.8
Hazard ratio 95% CI p Value
SFC vs placebo 0.83 0.70 to 0.98 0.031
Salmeterol vs placebo 0.96 0.82 to 1.13 0.629
FP vs placebo 1.00 0.85 to 1.18 0.994
CV SAEs
Patients with CV SAE, n (%) 176 (11) 168 (11) 180 (12) 160 (10)
Rate* of CV SAE (no of events) 75 (245) 66 (234) 66 (236) 57 (209)
Probabilityyof CV SAE by
3 years, %
15.4 13.6 14.7 12.5
Hazard ratio 95% CI p Value
SFC vs placebo 0.81 0.65 to 1.00 0.046
SAL vs placebo 0.89 0.72 to 1.10 0.268
FP vs placebo 0.94 0.76 to 1.15 0.542
*Rate¼number of events per 1000 treatment years.
yKaplaneMeier probability.
AEs, adverse events; CV, cardiovascular; FP, fluticasone propionate; SAEs, serious adverse
events; SFC, salmeterol/fluticasone propionate combination.
722 Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
but there was a similar pattern across treatment groups. Seven
percent of patients had a history of previous MI. In this group
the cardiovascular event rate, as would be expected, was higher.
Again, no trend was seen for more AEs in those patients rand-
omised to treatment with SAL in combination with FP.
However, the data for SAL alone are inconclusive, possibly due
to the small sample size in this smaller subgroup of patients.
Unlike other COPD studies, TORCH developed a rigorous
methodology for determining the likely cause of death which
was adjudicated by an expert panel blinded to the study medi-
cation.
18
Moreover, there was effectively complete follow-up of
the vital status of all patients 3 years after randomisation. In
this data set, the patients randomised to LABA alone had the
lowest rate of cardiovascular death while those who received
placebo had the largest number of events. The number of on-
treatment deaths, analogous to data included in earlier COPD
studies, showed a similar pattern across treatments.
A range of predictable factors increases the cardiovascular event
rate including higher age, a history of previous cardiac disease and
worse lung function. None of these factors interacted with
treatment to identify a specicat-riskgroup. Somewhat
surprisingly we saw no difference in event rate between current
and ex-smokers, which may reect our study entry criteria or
possibly the nature of cardiovascular disease in patients with
COPD. We did not control for the use of inhaled anticholinergic
drugs although, when used, this was predominantly ipratropium
as tiotropium was not available in most countries until
TORCH was nearing completion. In those patients treated with
an anticholinergic agent, there was a suggestion of a somewhat
higher cardiovascular event rate but these patients also had lower
baseline lung function. The association seen is therefore likely to
represent the confounding inuence of disease severity, an issue
that has made interpretation of previous studies of COPD
therapy particularly difcult. Patients in GOLD stage 4 receiving
salmeterol appeared to have more cardiac events, although the
Table 5 KaplaneMeier probability of a cardiovascular adverse event
by 3 years by subgroups
N
Placebo
(n[1544)
Salmeterol
(n[1542)
FP
(n[1552)
SFC
(n[546)
Age
<55 706 13.2 12.5 13.4 13.4
55e64 1988 20.2 18.8 20.7 17.4
65e74 2706 26.6 24.8 26.9 22.1
$75 784 39.2 34.9 35.4 32.9
Sex
Male 4684 24.4 23.0 24.0 20.7*
Female 1500 23.4 21.9 25.3 21.0
Smoking status
Current 2654 22.2 22.0 22.4 20.5
Former 3530 25.8 23.3 25.9 20.9*
Baseline FEV
1
<30% 945 25.8 32.1 26.7 23.5
30e<50% 3056 25.7 20.7 25.4 23.4
$50% 2183 21.7 21.7 21.8 16.2*
Baseline CV treatment
Yes 2530 33.5 30.3 30.4 27.9*
No 3654 18.3 17.6 20.2 15.4
Prior MI
Yes 419 49.1 54.4 41.0 44.1
No 5762 22.2 20.2 23.3 19.1
Baseline short-acting anticholinergics
Yes 3715 25.2 24.0 25.6 22.0
No 2469 22.7 21.0 22.4 18.9
*p<0.05 versus placebo.
CV, cardiovascular; FEV
1
, forced expiratory volume in 1 s; FP, fluticasone propionate;
MI, myocardial infarction; SFC, salmeterol/fluticasone propionate combination.
Table 4 Ischaemic cardiovascular AEs
Placebo
(n [1544)
Salmeterol
(n [1542)
FP
(n [1552)
SFC
(n [1546)
Patients with ischaemic
CV AE, n (%)
166 (11) 166 (11) 167 (11) 144 (9)
Rate* of ischaemic CV AE
(no of events)
68 (224) 70 (240) 62 (222) 54 (199)
Probabilityyof ischaemic
CV AE by 3 years, %
14.6 13.4 13.8 11.3
Hazard ratio 95% CI p Value
SFC vs placebo 0.76 0.61 to 0.95 0.016
Salmeterol vs
placebo
0.93 0.75 to 1.16 0.531
FP vs placebo 0.93 0.75 to 1.15 0.477
*Rate¼number of events per 1000 treatment years.
yKaplaneMeier probability.
AEs, adverse events; CV, cardiovascular; FP, fluticasone propionate;
SFC, salmeterol/fluticasone propionate combination.
Figure 3 Cumulative incidence of all cardiovascular adverse events in
patients who (A) did not receive or (B) did receive cardiovascular
medication (listed in table 1) in the 12 months prior to screening. Full
details of the data presented in this figure including statistical testing are
shown in table 5. Vertical bars represent standard errors. AE, adverse
event; CV, cardiovascular; FP, fluticasone propionate; SAL, salmeterol;
SFC, salmeterol/fluticasone propionate combination.
Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077 723
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
differences between treatment groups were not signicant. This
nding is at odds with the lower than average reported incidence
of events with salmeterol in GOLD stage 3 and probably reects
the lower sample size in the GOLD stage 4 population with
correspondingly widened condence intervals for these data.
Our study has strengths and some limitations. We monitored
our patients regularly throughout the 3-year study, but events
were self-reported rather than being in response to a prede-
termined diagnostic list. We did not undertake ECG or echo-
cardiographic evaluations at the study outset and there was no
requirement to provide objective documentation of the nature of
any new cardiovascular episode. However, we did include
patients with a history of cardiovascular disease provided it was
not thought that this was the main cause for their symptoms or
that they were likely to die from this during the study. This is an
important difference from earlier studies where more restrictive
eligibility criteria were applied. As in other large COPD studies,
for example UPLIFT
21
and TRISTAN,
22
patients randomised to
placebo tended to withdraw more frequently than those rand-
omised to active therapy, probably reecting their deteriorating
condition. Thus, patients continuing in the placebo arm of our
on-treatment analysis represent a relatively tter group of
patients with COPD. Despite this, we saw no suggestion that
patients on treatment were more likely to experience adverse
cardiovascular problems. Finally, we lack data about whether the
use of these agents increases the risk of cardiovascular events
during an acute exacerbation. Recent literature reviews have
failed to report any association between use of high-dose
b
agonists and risk of arrhythmias in this setting.
23
For some of our data the SFC combination appeared to be
associated with important reductions in the incidence of adverse
cardiovascular events. Although this difference may simply have
been due to chance, other biologically plausible mechanisms
exist which can account for this effect. It has been suggested
that inammation occurring in COPD might directly promote
vascular damage
24
and this may be reduced when airway
inammation is decreased, as has been demonstrated with
a LABA/inhaled steroid combination.
25
Our data cannot address
this hypothesis, but this concept is supported by recent obser-
vations from a large database study that patients who used
inhaled corticosteroids were less likely to experience cardiovas-
cular deaths than those who received bronchodilators alone.
12
This is consistent with the general conclusions of the TORCH
study that there was a reasonable (but not conclusive proba-
bility that combination treatment with SAL plus FP prolongs
life in COPD. An alternative explanation for the effect of SFC on
cardiovascular outcomes may be its relative efciency in
preventing exacerbations of COPD which are associated with
elevations in troponin T and a raised cardiac infarction injury
score, at least in hospitalised patients.
26 27
Further studies will
be needed to address whether these potentially important
mechanisms best explain the observed data.
In summary, in this large prospectively collected dataset, the
occurrence of new cardiovascular AEs was no more frequent in
patients treated with a LABA than in those treated with placebo.
In addition, we saw some evidence that the combination of
a LABA and an inhaled corticosteroid might offer a degree of
cardioprotection. These data from patients with moderate to
severe COPD provide reassurance that our current use of inhaled
LABA therapies is not harmful to cardiovascular health.
Acknowledgements The authors acknowledge medical writing support from David
Cutler, a professional medical writer with Gardiner-Caldwell Communications, in the
preparation of this manuscript; this support was funded by GlaxoSmithKline.
Funding This work was supported by GlaxoSmithKline.
Competing interests PMAC has received consulting fees from AstraZeneca,
GlaxoSmithKline, Nycomed and Pfizer; speaking fees from GlaxoSmithKline and
Nycomed; and grant support from Boehringer-Ingelheim and GlaxoSmithKline. JAA,
CC, LRW and JCY are employed by and hold stock in GlaxoSmithKline. BC has
received consulting fees from Altana, AstraZeneca, Boehringer-Ingelheim and
GlaxoSmithKline; speaking fees from Altana, AstraZeneca, Boehringer-Ingelheim and
GlaxoSmithKline; and grant support from Boehringer-Ingelheim and GlaxoSmithKline.
GTF has received consulting fees from Boehringer-Ingelheim, GlaxoSmithKline,
Novartis and Schering Plough; speaking fees from Boehringer-Ingelheim,
GlaxoSmithKline and Pfizer; and grant support from Altana, Boehringer-Ingelheim,
Emphasys Medical Inc, Forrest, GlaxoSmithKline, Mannkind Corporation and Novartis.
CJ has received consulting fees from Altana, AstraZeneca, Boehringer-Ingelheim and
GlaxoSmithKline; speaking fees from Altana, AstraZeneca, Boehringer-Ingelheim,
GlaxoSmithKline and Novartis; and grant support from GlaxoSmithKline. PWJ has
received consulting fees from AstraZeneca, GlaxoSmithKline, Novartis and Roche;
speaking fees from AstraZeneca and GlaxoSmithKline; and grant support from
Boehringer-Ingelheim and GlaxoSmithKline. JV has received consulting fees from
AstraZeneca, Boehringer-Ingelheim, GlaxoSmithKline, Hoffman-LaRoche and
Nycomed; speaking fees from AstraZeneca, Boehringer-Ingelheim and
GlaxoSmithKline; and grant support from GlaxoSmithKline.
Ethics approval The study was approved by local ethics review committees and was
conducted in accordance with the Declaration of Helsinki and Good Clinical Practice
guidelines. All patients gave written informed consent.
Provenance and peer review Not commissioned; externally peer reviewed.
Figure 4 Cumulative incidence of all cardiovascular adverse events in
patients who (A) had not experienced or (B) had experienced
a myocardial infarction prior to study entry. Full details of the data
presented in this figure including statistical testing are shown in table 5.
Vertical bars represent standard errors. AE, adverse event; FP,
fluticasone propionate; MI, myocardial infarction; SAL, salmeterol;
SFC, salmeterol/fluticasone propionate combination.
724 Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
REFERENCES
1. Viegi G, Pistelli F, Sherrill DL, et al. Definition, epidemiology and natural history of
COPD. Eur Respir J 2007;30:993e1013.
2. Lopez AD, Shibuya K, Rao C, et al. Chronic obstructive pulmonary disease: current
burden and future projections. Eur Respir J 2006;27:397e412.
3. Sin DD, Man SF. Chronic obstructive pulmonary disease as a risk factor for
cardiovascular morbidity and mortality. Proc Am Thorac Soc 2005;2:8e11.
4. Anthonisen NR, Skeans MA, Wise RA, et al. Lung Health Study Research Group.
The effects of a smoking cessation intervention on 14.5-year mortality: a randomized
clinical trial. Ann Intern Med 2005;142:233e9.
5. Keistinen T, Tuuponen T, Kivela SL. Survival experience of the population needing
hospital treatment for asthma or COPD at age 50e54 years. Respir Med
1998;92:568e72.
6. Curkendall SM, Lanes S, de Luise C, et al. Chronic obstructive pulmonary disease
severity and cardiovascular outcomes. Eur J Epidemiol 2006;21:803e13.
7. Johnston AK, Mannino DM, Hagan GW, et al. Relationship between lung function
impairment and incidence or recurrence of cardiovascular events in a middle-aged
cohort. Thorax 2008;63:599e605.
8. Hole DJ, Watt GC, Davey-Smith G, et al. Impaired lung function and mortality risk in
men and women: findings from the Renfrew and Paisley prospective population
study. BMJ 1996;313:711e15.
9. Curkendall SM, DeLuise C, Jones JK, et al. Cardiovascular disease in patients with
chronic obstructive pulmonary disease, Saskatchewan Canada cardiovascular
disease in COPD patients. Ann Epidemiol 2006;16:63e70.
10. Singh S, Loke YK, Furberg CD. Inhaled anticholinergics and risk of major adverse
cardiovascular events in patients with chronic obstructive pulmonary disease:
a systematic review and meta-analysis. JAMA 2008;300:1439e50.
11. Macie C, Wooldrage K, Manfreda J, et al. Cardiovascular morbidity and the use of
inhaled bronchodilators. Int J Chron Obstruct Pulmon Dis 2008;3:163e9.
12. Lee TA, Pickard AS, Au DH, et al. Risk for death associated with medications for
recently diagnosed chronic obstructive pulmonary disease. Ann Intern Med
2008;149:380e90.
13. Cazzola M, Matera MG, Donner CF. Inhaled beta2-adrenoceptor agonists:
cardiovascular safety in patients with obstructive lung disease. Drugs
2005;65:1595e610.
14. Salpeter SR, Ormiston TM, Salpeter EE. Cardiovascular effects of beta-agonists in
patients with asthma and COPD: a meta-analysis. Chest 2004;125:2309e21.
15. Calverley PM, Anderson JA, Celli B, et al. TORCH investigators. Salmeterol and
fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl
J Med 2007;356:775e89.
16. Ferguson GT, Calverley PMA, Anderson JA, et al. Incidence of cardiac events in
COPD patients in the TORCH study [abstract]. Am J Respir Crit Care Med
2008;177:A962.
17. Vestbo J. Torch Study Group. The TORCH (towards a revolution in COPD health)
survival study protocol. Eur Respir J 2004;24:206e10.
18. McGarvey LP, John M, Anderson JA, et al. TORCH Clinical Endpoint Committee.
Ascertainment of cause-specific mortality in COPD: operations of the TORCH Clinical
Endpoint Committee. Thorax 2007;62:411e15.
19. Fabbri LM, Rabe KF. From COPD to chronic systemic inflammatory syndrome?
Lancet 2007;370:797e9.
20. Mills NL, Miller JJ, Anand A, et al. Increased arterial stiffness in patients with
chronic obstructive pulmonary disease: a mechanism for increased cardiovascular
risk. Thorax 2008;63:306e11.
21. Tashkin DP, Celli B, Senn S, et al. Uplift Study Investigators. A 4-year trial of
tiotropium in chronic obstructive pulmonary disease. N Engl J Med
2008;359:1543e54.
22. Calverley P, Pauwels R, Vestbo J, et al. TRial of Inhaled STeroids ANd long-acting
beta2 agonists study group. Combined salmeterol and fluticasone in the treatment of
chronic obstructive pulmonary disease: a randomised controlled trial. Lancet
2003;361:449e56.
23. Maak CA, Tabas JA, McClintock DE. Should acute treatment with inhaled beta
agonists be withheld from patients with dyspnea who may have heart failure?
J Emerg Med. Published Online First: 2008 Jun 20.
24. Calverley PM, Scott S. Is airway inflammation in chronic obstructive pulmonary
disease (COPD) a risk factor for cardiovascular events? COPD 2006;3:233e42.
25. Barnes NC, Qiu YS, Pavord ID, et al. SCO30005 Study Group. Antiinflammatory
effects of salmeterol/fluticasone propionate in chronic obstructive lung disease.
Am J Respir Crit Care Med 2006;173:736e43.
26. Brekke PH, Omland T, Smith P, et al. Underdiagnosis of myocardial infarction in
COPD: Cardiac Infarction Injury Score (CIIS) in patients hospitalised for COPD
exacerbation. Respir Med 2008;102:1243e7.
27. Brekke PH, Omland T, Holmedal SH, et al. Troponin T elevation and long-term
mortality after chronic obstructive pulmonary disease exacerbation. Eur Respir J
2008;31:563e70.
Thorax 2010;65:719e725. doi:10.1136/thx.2010.136077 725
Chronic obstructive pulmonary disease
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
TORCH Study results
Cardiovascular events in patients with COPD:
Julie C Yates and Jørgen Vestbo Willits,Ferguson, Christine Jenkins, Paul W Jones, Courtney Crim, Lisa R
Peter M A Calverley, Julie A Anderson, Bartolome Celli, Gary T
doi: 10.1136/thx.2010.136077
2010 65: 719-725 Thorax
http://thorax.bmj.com/content/65/8/719
Updated information and services can be found at:
These include:
Material
Supplementary http://thorax.bmj.com/content/suppl/2010/08/06/65.8.719.DC1.html
Supplementary material can be found at:
References #BIBLhttp://thorax.bmj.com/content/65/8/719
This article cites 26 articles, 8 of which you can access for free at:
service
Email alerting box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in the
Collections
Topic Articles on similar topics can be found in the following collections
(113)Ischaemic heart disease (493)Drugs: respiratory system
Notes
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
group.bmj.com on October 18, 2015 - Published by http://thorax.bmj.com/Downloaded from
... Following a pos-thoc analysis of the TORCH trial showing a significant reduction of cardiovascular mortality in the ICS/LABA combination [86] the SUMMIT [83] study was designed. The study was conducted in 1368 centers and included more than 16000 COPD patients who either suffered from cardiovascular comorbidities or were at increased risk for cardiovascular death. ...
Article
Full-text available
COPD is a major healthcare problem and cause of mortality worldwide. COPD patients at increased mortality risk are those that are more symptomatic, have lower lung function and lower DLCO, have decreased exercise capacity, belong to the emphysematous phenotype and those who have concomitant bronchiectasis. Also mortality risk seems to be greater in patients who experience COPD exacerbations and in those who suffer from concomitant cardiovascular and/or metabolic diseases. To predict the risk of death in COPD patients several composite scores have been created using different parameters. In the previous years, large studies (also called Mega-Trials) have evaluated the efficacy of different therapies on COPD mortality, but until recently only non-pharmaceutical interventions have proven to be effective. However, recent studies on fixed combinations of triple therapy (long-acting beta agonists, long-acting muscarinic antagonists and inhaled corticosteroids) have provided encouraging results, showing for the first time a reduction in mortality compared to dual therapies. The aim of the present review is to summarize available data regarding mortality risk in COPD patients and to describe pharmacologic therapies that have shown effectiveness in reducing mortality.
... According to clinical guidelines, COPD should be treated in patients with HF because there is no direct evidence that this respiratory disease should be treated differently in the presence of HF [34]. This statement is based on long-term study findings in patients with HF and comorbid COPD [43][44][45]. Some studies, but not all, have suggested that beta-2 agonists may be detrimental to patients with left ventricular dysfunction. ...
Article
Full-text available
The convergence of chronic obstructive pulmonary disease (COPD) and heart failure (HF) is a prevalent yet often overlooked medical scenario. This coexistence poses diagnostic challenges due to symptom similarities. This comprehensive review extensively examines the impact of COPD and HF on pharmacological management. Furthermore, the concurrent presence of these conditions amplifies both mortality rates and societal financial strain. Addressing these intertwined ailments necessitates a multidisciplinary approach. Within this review, we delve into the foundational mechanisms, diagnostic intricacies, and available management choices for these closely related conditions.
... This impact may be especially relevant for individuals with underlying cardiac disorders, albeit those with heart failure frequently have bronchoconstriction that is reversible with inhaled β 2 -agonists. The resulting decrease in breathing effort might reduce cardiac workload even more [93][94][95][96][97][98]. Furthermore, there is evidence that in patients with pulmonary hyperinflation, which is frequent in both asthma and COPD, dual bronchodilation improves regional ventilation and blood flow in the pulmonary microcirculation. ...
Article
Full-text available
Pharmacological management of airway obstructive diseases is a fast-evolving field. Several advances in unravelling disease mechanisms as well as intracellular and molecular pathways of drug action have been accomplished. While the clinical translation and implementation of in vitro results to the bedside remains challenging, advances in comprehending the mechanisms of respiratory medication are expected to assist clinicians and scientists in identifying meaningful read-outs and designing clinical studies. This European Respiratory Society Research Seminar, held in Naples, Italy, 5–6 May 2022, focused on current and future developments of the drugs used to treat asthma and COPD; on mechanisms of drug action, steroid resistance, comorbidities and drug interactions; on prognostic and therapeutic biomarkers; on developing novel drug targets based on tissue remodelling and regeneration; and on pharmacogenomics and emerging biosimilars. Related European Medicines Agency regulations are also discussed, as well as the seminar's position on the above aspects.
... 42 Nevertheless, a post hoc analysis of the Toward a Revolution in COPD Health (TORCH) study concluded that although a history of myocardial infarction doubled the risk of adverse CVEs, the development of new CVEs was not more common in patients who received LABA than in placebo-treated individuals. 43 Dual bronchodilation. The development of LAMA/LABA combinations was undoubtedly a significant advancement in the treatment of COPD patients, 44 and such a therapeutic approach is currently the cornerstone of recommendations in the Global Initiative for Chronic ...
Article
Full-text available
Chronic airway diseases (CAD), mainly asthma and chronic obstructive pulmonary disease (COPD), are frequently associated with different comorbidities. Among them, cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) pose problems for the simultaneous treatment of CAD and comorbidity. Indeed, there is evidence that some drugs used to treat CAD negatively affect comorbidity, and, conversely, some drugs used to treat comorbidity may aggravate CAD. However, there is also growing evidence of some beneficial effects of CAD drugs on comorbidities and, conversely, of the ability of some of those used to treat comorbidity to reduce the severity of lung disease. In this narrative review, we first describe the potential cardiovascular risks and benefits for patients using drugs to treat CAD and the potential lung risks and benefits for patients using drugs to treat CVD. Then, we illustrate the possible negative and positive effects on T2DM of drugs used to treat CAD and the potential negative and positive impact on CAD of drugs used to treat T2DM. The frequency with which CAD and CVD or T2DM are associated requires not only considering the effect that drugs used for one disease condition may have on the other but also providing an opportunity to develop therapies that simultaneously favorably impact both diseases.
... A classic example is chronic obstructive pulmonary disease (COPD) where the most frequent comorbidities are cardiovascular. 3,4 Similarly, patients with cardiovascular diseases, especially ischemic, usually demonstrate signs of respiratory impairment, which may need to be investigated by pulmonary function tests (spirometry). [5][6][7] The main mechanical defect in the most common chronic lung diseases (COPD and asthma) is lung hyperinflation (LH). ...
Article
Full-text available
In this review, we present the effects of lung hyperinflation on the cardiovascular system (CVS) and the beneficial outcomes of different deflation treatment modalities. We discuss the effects of long-acting bronchodilator drugs, medical and surgical lung volume reduction on the performance of the CVS. Although there is a small number of studies investigating lung deflation and the CVS, the short-term improvement in heart function was clearly demonstrated. However, more studies, with longer duration, are needed to verify these significant beneficial effects of deflation of the lungs on the CVS. Dynamic hyperinflation during exercise could be a research model to investigate further the effects of lung hyperinflation and/or deflation on the CVS.
Article
Создание руководства поддержано Советом по терапевтическим наукам отделения клинической медицины Российской академии наук.
Article
Background In the majority of current therapeutic regimens for chronic obstructive pulmonary disease (COPD), bronchodilators are coupled with inhaled corticosteroids (ICS) to lower the inflammatory response and improve symptoms. This study aims to evaluate the safety of ICS in the treatment of COPD. Methods Randomized controlled trials related to ICS for COPD that were eligible up to 1 June 2023 were searched in PubMed, EMBASE, and Cochrane. We searched and screened eligible studies for the occurrence of total adverse events, cardiovascular events, upper respiratory tract infections (URTI), pneumonia, oral Candida infections, and musculoskeletal disorders, and finally analyzed them by Review Manager 5.4.1. Results The results showed that ICS increased the incidence of adverse reactions in COPD patients (RR = 1.06, 95% CI: 1.03–1.10, P = .0004); ICS treatment did not increase the risk of cardiovascular events in COPD patients (RR = 0.95, 95% CI: 0.88–1.02, P = .14); ICS increased the incidence of URTI in COPD patients (RR = 1.29, 95% CI: 1.02–1.62, P = .03); ICS increased the incidence of pneumonia in patients with COPD (RR = 2.09, 95% CI: 1.63–2.69, P < .00001); ICS treatment significantly increased the incidence of oral Candida in patients with COPD (RR = 2.96, 95% CI: 1.99–4.41, P < .00001); ICS increased the incidence of musculoskeletal disorders in patients with COPD (RR = 2.87, 95% CI: 1.51–5.45, P = .001). Conclusion ICS does not increase the risk of cardiovascular events in patients with COPD, but it does increase the risk of URTI, pneumonia, oral Candida infections, and musculoskeletal disorders in patients with COPD.
Article
The initial alterations of chronic obstructive pulmonary disease (COPD) involve the small airways. Small airway disease (SAD) is related to lung hyperinflation and air trapping. Several lung function tests may detect the presence of SAD, namely forced mid-expiratory flows, residual volume (RV), RV/total lung capacity (TLC) ratio, functional residual capacity, airway resistances obtained with body-plethysmography and oscillometry, and the single-breath nitrogen washout test. Additionally, high-resolution computed tomography can detect SAD. In addition to SAD, COPD is related to cardiovascular disease (CVD) such as heart failure, peripheral vascular disease, and ischemic heart disease. No studies have assessed the relationship between CVD, COPD, and SAD. Therefore, the main objective of the Assessing the Relationship between Cardiovascular and small Airway Disease and Acute events in COPD (ARCADIA) study is to assess the risk of CVD in COPD patients according to SAD in a real-life setting. The correlation between CVD, mortality, and acute exacerbation of COPD (AECOPD) is also evaluated. ARCADIA is a 52-week prospective, multicentre, pilot, observational, cohort study conducted in ≥22 pulmonary centres in Italy and that enrols ≥500 COPD patients, regardless of disease severity (protocol registration: ISRCTN49392136). SAD is evaluated at baseline, after that CVD, mortality, and AECOPD are recorded at 6 and 12 months. Bayesian inference is used to quantify the risk and correlation of the investigated outcomes in COPD patients according to SAD. The ARCADIA study provides relevant findings in the daily clinical management of COPD patients.
Article
Background: The role of inhaled corticosteroids (ICS) in chronic obstructive pulmonary disease (COPD) has been the subject of much uncertainty. COPD clinical guidelines currently recommend selective use of ICS. ICS are not recommended as monotherapy for people with COPD, and are only given in combination with long-acting bronchodilators due to greater efficacy of combination therapy. Incorporating and critiquing newly published placebo-controlled trials into the monotherapy evidence base may help to resolve ongoing uncertainties and conflicting findings about their role in this population. Objectives: To evaluate the benefits and harms of inhaled corticosteroids, used as monotherapy versus placebo, in people with stable COPD, in terms of objective and subjective outcomes. Search methods: We used standard, extensive Cochrane search methods. The latest search date was October 2022. Selection criteria: We included randomised trials comparing any dose of any type of ICS, given as monotherapy, with a placebo control in people with stable COPD. We excluded studies of less than 12 weeks' duration and studies of populations with known bronchial hyper-responsiveness (BHR) or bronchodilator reversibility. Data collection and analysis: We used standard Cochrane methods. Our a priori primary outcomes were 1. exacerbations of COPD and 2. quality of life. Our secondary outcomes were 3. all-cause mortality, 4. lung function (rate of decline of forced expiratory volume in one second (FEV1)), 5. rescue bronchodilator use, 6. exercise capacity, 7. pneumonia and 8. adverse events including pneumonia. ]. We used GRADE to assess certainty of evidence. Main results: Thirty-six primary studies with 23,139 participants met the inclusion criteria. Mean age ranged from 52 to 67 years, and females were 0% to 46% of participants. Studies recruited across the severities of COPD. Seventeen studies were of duration longer than three months and up to six months and 19 studies were of duration longer than six months. We judged the overall risk of bias as low. Long-term (more than six months) use of ICS as monotherapy reduced the mean rate of exacerbations in those studies where pooling of data was possible (generic inverse variance analysis: rate ratio 0.88 exacerbations per participant per year, 95% confidence interval (CI) 0.82 to 0.94; I2 = 48%, 5 studies, 10,097 participants; moderate-certainty evidence; pooled means analysis: mean difference (MD) -0.05 exacerbations per participant per year, 95% CI -0.07 to -0.02; I2 = 78%, 5 studies, 10,316 participants; moderate-certainty evidence). ICS slowed the rate of decline in quality of life, as measured by the St George's Respiratory Questionnaire (MD -1.22 units/year, 95% CI -1.83 to -0.60; I2 = 0%; 5 studies, 2507 participants; moderate-certainty evidence; minimal clinically importance difference 4 points). There was no evidence of a difference in all-cause mortality in people with COPD (odds ratio (OR) 0.94, 95% CI 0.84 to 1.07; I2 = 0%; 10 studies, 16,636 participants; moderate-certainty evidence). Long-term use of ICS reduced the rate of decline in FEV1 in people with COPD (generic inverse variance analysis: MD 6.31 mL/year benefit, 95% CI 1.76 to 10.85; I2 = 0%; 6 studies, 9829 participants; moderate-certainty evidence; pooled means analysis: 7.28 mL/year, 95% CI 3.21 to 11.35; I2 = 0%; 6 studies, 12,502 participants; moderate-certainty evidence). Adverse events: in the long-term studies, the rate of pneumonia was increased in the ICS group, compared to placebo, in studies that reported pneumonia as an adverse event (OR 1.38, 95% CI 1.02 to 1.88; I2 = 55%; 9 studies, 14,831 participants; low-certainty evidence). There was an increased risk of oropharyngeal candidiasis (OR 2.66, 95% CI 1.91 to 3.68; 5547 participants) and hoarseness (OR 1.98, 95% CI 1.44 to 2.74; 3523 participants). The long-term studies that measured bone effects generally showed no major effect on fractures or bone mineral density over three years. We downgraded the certainty of evidence to moderate for imprecision and low for imprecision and inconsistency. Authors' conclusions: This systematic review updates the evidence base for ICS monotherapy with newly published trials to aid the ongoing assessment of their role for people with COPD. Use of ICS alone for COPD likely results in a reduction of exacerbation rates of clinical relevance, probably results in a reduction in the rate of decline of FEV1 of uncertain clinical relevance and likely results in a small improvement in health-related quality of life not meeting the threshold for a minimally clinically important difference. These potential benefits should be weighed up against adverse events (likely to increase local oropharyngeal adverse effects and may increase the risk of pneumonia) and probably no reduction in mortality. Though not recommended as monotherapy, the probable benefits of ICS highlighted in this review support their continued consideration in combination with long-acting bronchodilators. Future research and evidence syntheses should be focused in that area.
Article
Full-text available
Background β-Adrenergic agonists exert physiologic effects that are the opposite of those of β-blockers. β-Blockers are known to reduce morbidity and mortality in patients with cardiac disease. β2-Agonist use in patients with obstructive airway disease has been associated with an increased risk for myocardial infarction, congestive heart failure, cardiac arrest, and acute cardiac death. Objectives To assess the cardiovascular safety of β2-agonist use in patients with obstructive airway disease, defined as asthma or COPD. Methods A meta-analysis of randomized placebo-controlled trials of β2-agonist treatment in patients with obstructive airway disease was performed, to evaluate the short-term effect on heart rate and potassium concentrations, and the long-term effect on adverse cardiovascular events. Longer duration trials were included in the analysis if they reported at least one adverse event. Adverse events included sinus and ventricular tachycardia, syncope, atrial fibrillation, congestive heart failure, myocardial infarction, cardiac arrest, or sudden death. Results Thirteen single-dose trials and 20 longer duration trials were included in the study. A single dose of β2-agonist increased the heart rate by 9.12 beats/min (95% confidence interval [CI], 5.32 to 12.92) and reduced the potassium concentration by 0.36 mmol/L (95% CI, 0.18 to 0.54), compared to placebo. For trials lasting from 3 days to 1 year, β2-agonist treatment significantly increased the risk for a cardiovascular event (relative risk [RR], 2.54; 95% CI, 1.59 to 4.05) compared to placebo. The RR for sinus tachycardia alone was 3.06 (95% CI, 1.70 to 5.50), and for all other events it was 1.66 (95% CI, 0.76 to 3.6). Conclusion β2-Agonist use in patients with obstructive airway disease increases the risk for adverse cardiovascular events. The initiation of treatment increases heart rate and reduces potassium concentrations compared to placebo. It could be through these mechanisms, and other effects of β-adrenergic stimulation, that β2-agonists may precipitate ischemia, congestive heart failure, arrhythmias, and sudden death.
Article
Full-text available
Christine Macie, Kate Wooldrage, Jure Manfreda, Nicholas AnthonisenDepartment of Medicine, University of Manitoba, Winnipeg, Manitoba, CanadaAbstract: We used the Manitoba Health database to examine the relationship between use of inhaled respiratory drugs in people with chronic obstructive respiratory diseases and cardiovascular hospitalizations from 1996 through 2000. The drugs examined were beta agonists [BA], ipratropium bromide IB, and inhaled steroids (ICS). End points were first hospitalizations for supraventricular tachycardia, myocardial infarction, heart failure or stroke. A nested case control analysis was employed comparing people with and without cardiovascular events. Cases and controls were matched for gender and age, and conditional logistic regression was used in multivariate analysis considering other respiratory drugs, respiratory diagnosis and visit frequency, non-respiratory, non-cardiac comorbidities, and receipt of drugs for cardiovascular disease. In univariate analyses, BA, IB and ICS were all associated with hospitalizations for cardiovascular disease, but in multivariate analyses ICS did not increase risk while both BA and IB did. There were interactions between respiratory and cardiac drugs receipt in that bronchodilator associated risks were higher in people not taking cardiac drugs; this was especially true for stroke. There were strong interactions with specific cardiac drugs; for example, both BA and IB substantially increased the risk of supraventricular tachycardia in patients not anti-arryhthmic agents, but not in the presence of such agents. We conclude that bronchodilator therapy for chronic obstructive diseases is associated with increased cardiovascular risk, especially in patients without previous cardiovascular diagnoses, and that this is unlikely due to the severity of the respiratory disease, since risk was not increased with ICS.Keywords: bronchodilator therapy, inhaled corticosteroids, nested case control study
Article
Full-text available
Inhaled anticholinergics (ipratropium bromide or tiotropium bromide) are widely used in patients with chronic obstructive pulmonary disease (COPD) but their effect on the risk of cardiovascular outcomes is unknown. To ascertain the cardiovascular risks of inhaled anticholinergics, including cardiovascular death, myocardial infarction (MI), and stroke. Systematic searches were conducted on March 19, 2008, of relevant articles in MEDLINE, the Cochrane Database of systematic reviews, regulatory authority Web sites in the United States and the United Kingdom, and manufacturers' trial registries with no date restrictions. Randomized controlled trials of any inhaled anticholinergic for treatment of COPD that had at least 30 days of treatment and reported on cardiovascular events. The primary outcome was a composite of cardiovascular death, MI, or stroke. The secondary outcome was all-cause mortality. Relative risks (RRs) were estimated using fixed-effects models and statistical heterogeneity was estimated with the I(2) statistic. After a detailed screening of 103 articles, 17 trials enrolling 13,645 [corrected] patients were analyzed. Follow-up duration ranged from 6 weeks to 5 years. Cardiovascular death, MI, or stroke occurred in 134 of 6984 [corrected] patients (1.9%) [corrected] receiving inhaled anticholinergics and 83 of 6661 [corrected] patients (1.2%) receiving control therapy (RR, 1.60 [corrected] [95% confidence interval {CI}, 1.22-2.10]; [corrected] P < .001, I(2) = 0%). Among individual components of the primary end point, inhaled anticholinergics significantly increased the risk of MI (RR, 1.52 [95% CI 1.04-2.22]; [corrected] P = .03, I(2) = 0%) and cardiovascular death (RR, 1.92 [95% CI, 1.23-3.00]; P = .004, [corrected] I(2) = 0%) without a statistically significant increase in the risk of stroke (RR, 1.46 [95% CI, 0.81-2.62]; P = .20, I(2) = 0%). All-cause mortality was reported in 146 [corrected] of the patients treated with inhaled anticholinergics (2.1%) and 108 [corrected] of the control patients (1.6%) (RR, 1.29 [95% CI, 1.00-1.65]; P = .05, I(2) = 0%) [corrected] A sensitivity analysis restricted to 6 [corrected] long-term trials (>6 months) confirmed the significantly increased risk of cardiovascular death, MI, or stroke (2.9% of patients treated with anticholinergics vs 1.8% of the control patients; RR, 1.73 [95% CI, 1.27-2.35]; [corrected] P < .001, I(2) = 0%). Inhaled anticholinergics are associated with a significantly increased risk of cardiovascular death, MI, or stroke among patients with COPD.
Article
Only long-term home oxygen therapy has been shown in randomised controlled trials to increase survival in chronic obstructive pulmonary disease (COPD). There have been no trials assessing the effect of inhaled corticosteroids and long-acting bronchodilators, alone or in combination, on mortality in patients with COPD, despite their known benefit in reducing symptoms and exacerbations. The "TOwards a Revolution in COPD Health" (TORCH) survival study is aiming to determine the impact of salmeterol/fluticasone propionate (SFC) combination and the individual components on the survival of COPD patients. TORCH is a multicentre, randomised, double-blind, parallel-group, placebo-controlled study. Approximately 6,200 patients with moderate-to-severe COPD were randomly assigned to b.i.d. treatment with either SFC (50/500 mug), fluticasone propionate (500 mug), salmeterol (50 mug) or placebo for 3 yrs. The primary end-point is all-cause mortality; secondary end-points are COPD morbidity relating to rate of exacerbations and health status, using the St George's Respiratory Questionnaire. Other end-points include other mortality and exacerbation end-points, requirement for long-term oxygen therapy, and clinic lung function. Safety end-points include adverse events, with additional information on bone fractures. The first patient was recruited in September 2000 and results should be available in 2006. This paper describes the "TOwards a Revolution in COPD Health" study and explains the rationale behind it.
Article
Previous studies showing that tiotropium improves multiple end points in patients with chronic obstructive pulmonary disease (COPD) led us to examine the long-term effects of tiotropium therapy. In this randomized, double-blind trial, we compared 4 years of therapy with either tiotropium or placebo in patients with COPD who were permitted to use all respiratory medications except inhaled anticholinergic drugs. The patients were at least 40 years of age, with a forced expiratory volume in 1 second (FEV(1)) of 70% or less after bronchodilation and a ratio of FEV(1) to forced vital capacity (FVC) of 70% or less. Coprimary end points were the rate of decline in the mean FEV(1) before and after bronchodilation beginning on day 30. Secondary end points included measures of FVC, changes in response on St. George's Respiratory Questionnaire (SGRQ), exacerbations of COPD, and mortality. Of a total of 5993 patients (mean age, 65+/-8 years) with a mean FEV(1) of 1.32+/-0.44 liters after bronchodilation (48% of predicted value), we randomly assigned 2987 to the tiotropium group and 3006 to the placebo group. Mean absolute improvements in FEV(1) in the tiotropium group were maintained throughout the trial (ranging from 87 to 103 ml before bronchodilation and from 47 to 65 ml after bronchodilation), as compared with the placebo group (P<0.001). After day 30, the differences between the two groups in the rate of decline in the mean FEV(1) before and after bronchodilation were not significant. The mean absolute total score on the SGRQ was improved (lower) in the tiotropium group, as compared with the placebo group, at each time point throughout the 4-year period (ranging from 2.3 to 3.3 units, P<0.001). At 4 years and 30 days, tiotropium was associated with a reduction in the risks of exacerbations, related hospitalizations, and respiratory failure. In patients with COPD, therapy with tiotropium was associated with improvements in lung function, quality of life, and exacerbations during a 4-year period but did not significantly reduce the rate of decline in FEV(1). (ClinicalTrials.gov number, NCT00144339.)
Article
Concerns exist regarding increased risk for mortality associated with some chronic obstructive pulmonary disease (COPD) medications. To examine the association between various respiratory medications and risk for death in veterans with newly diagnosed COPD. Nested case-control study in a cohort identified between 1 October 1999 and 30 September 2003 and followed through 30 September 2004 by using National Veterans Affairs inpatient, outpatient, pharmacy, and mortality databases; Centers for Medicare & Medicaid Services databases; and National Death Index Plus data. Cause of death was ascertained for a random sample of 40% of those who died during follow-up. Case patients were categorized on the basis of all-cause, respiratory, or cardiovascular death. Mortality risk associated with medications was assessed by using conditional logistic regression adjusted for comorbid conditions, health care use, and markers of COPD severity. U.S. Veterans Health Administration health care system. 32 130 case patients and 320 501 control participants in the all-cause mortality analysis. Of 11 897 patients with cause-of-death data, 2405 case patients had respiratory deaths and 3159 case patients had cardiovascular deaths. All-cause mortality; respiratory and cardiovascular deaths; and exposure to COPD medications, inhaled corticosteroids, ipratropium, long-acting beta-agonists, and theophylline in the 6 months preceding death. Adjusted odds ratios (ORs) for all-cause mortality were 0.80 (95% CI, 0.78 to 0.83) for inhaled corticosteroids, 1.11 (CI, 1.08 to 1.15) for ipratropium, 0.92 (CI, 0.88 to 0.96) for long-acting beta-agonists, and 1.05 (CI, 0.99 to 1.10) for theophylline. Ipratropium was associated with increased cardiovascular deaths (OR, 1.34 [CI, 1.22 to 1.47]), whereas inhaled corticosteroids were associated with reduced risk for cardiovascular death (OR, 0.80 [CI, 0.72 to 0.88]). Results were consistent across sensitivity analyses. Current smoking status and lung function were not measured. Misclassification of cause-specific mortality is unknown. The possible association between ipratropium and elevated risk for all-cause and cardiovascular death needs further study.
Article
To assess the relation between forced expiratory volume in one second (FEV1) and subsequent mortality. Prospective general population study. Renfrew and Paisley, Scotland. 7058 men and 8353 women aged 45-64 years at baseline screening in 1972-6. Mortality from all causes, ischaemic heart disease, cancer, hung and other cancers, stroke, respiratory disease, and other causes of death after 15 years of follow up. 2545 men and 1894 women died during the follow up period. Significant trends of increasing risk with diminishing FEV1 are apparent for both sexes for all the causes of death examined after adjustment for age, cigarette smoking, diastolic blood pressure, cholesterol concentration, body mass index, and social class. The relative hazard ratios for all cause mortality for subjects in the lowest fifth of the FEV1 distribution were 1.92 (95% confidence interval 1.68 to 2.20) for men and 1.89 (1.63 to 2.20) for women. Corresponding relative hazard ratios were 1.56 (1.26 to 1.92) and 1.88 (1.44 to 2.47) for ischaemic heart disease, 2.53 (1.69 to 3.79) and 4.37 (1.84 to 10.42) for lung cancer, and 1.66 (1.07 to 2.59) and 1.65 (1.09 to 2.49) for stroke. Reduced FEV1 was also associated with an increased risk for each cause of death examined except cancer for lifelong nonsmokers. Impaired lung function is a major clinical indicator of mortality risk in men and women for a wide range of diseases. The use of FEV1 as part of any health assessment of middle aged patients should be considered. Smokers with reduced FEV1 should form a priority group for targeted advice to stop smoking.
Article
The aim was to evaluate the differences of mortality among asthma and COPD patients on the basis of the first period of hospitalization of these diseases. A total of 576,916 treatment periods for asthma and COPD between 1972 and 1992 were identified in the discharge register maintained by the National Research and Development Centre for Welfare and Health. Patients aged 50-54 years first treated in hospital in 1977 or later were analysed. There were 6655 new asthma patients of this age, 2727 new COPD patients and 701 new patients in a mixed group (with both diagnoses). Mortality up to the end of 1993 was analysed based on mortality and cause of death data provided by the Central Statistical Office of Finland. Estimated cumulative survival after 10 yr was higher among the asthma patients (83.5% for men and 93.2% for women) and lower among the COPD patients (60.1% and 78.0%) and in the mixed group (62.5% and 74.4%). The main cause of death among the asthmatics was asthma in 12.1% of cases, that among the COPD patients was COPD in 22.1% of cases and that in the mixed group was one or other of these diseases in 39.1% of cases. The prognosis for COPD patients aged 50-54 years requiring hospital treatment is poor. Combination of the data available from the cause of death and hospital discharge registers indicates that obstructive pulmonary diseases may well be of more significance from a public health point of view than the mortality statistics would lead us to believe. The very first hospitalization for COPD calls for a thorough evaluation of the prospects for active treatment and prevention.
Article
Inhaled long-acting beta2 agonists improve lung function and health status in symptomatic chronic obstructive pulmonary disease (COPD), whereas inhaled corticosteroids reduce the frequency of acute episodes of symptom exacerbation and delay deterioration in health status. We postulated that a combination of these treatments would be better than each component used alone. 1465 patients with COPD were recruited from outpatient departments in 25 countries. They were treated in a randomised, double-blind, parallel-group, placebo-controlled study with either 50 microg salmeterol twice daily (n=372), 500 microg fluticasone twice daily (n=374), 50 microg salmeterol and 500 microg fluticasone twice daily (n=358), or placebo (n=361) for 12 months. The primary outcome was the pretreatment forced expiratory volume in 1s (FEV1) after 12 months treatment' and after patients had abstained from all bronchodilators for at least 6h and from study medication for at least 12h. Secondary outcomes were other lung function measurements, symptoms and rescue treatment use, the number of exacerbations, patient withdrawals, and disease-specific health status. We assessed adverse events, serum cortisol concentrations, skin bruising, and electrocardiograms. Analysis was as predefined in the study protocol. All active treatments improved lung function, symptoms, and health status and reduced use of rescue medication and frequency of exacerbations. Combination therapy improved pretreatment FEV1 significantly more than did placebo (treatment difference 133 mL, 95% CI 105-161, p<0.0001), salmeterol (73 mL, 46-101, p<0.0001), or fluticasone alone (95 mL, 67-122, p<0.0001). Combination treatment produced a clinically significant improvement in health status and the greatest reduction in daily symptoms. All treatments were well tolerated with no difference in the frequency of adverse events, bruising, or clinically significant falls in serum cortisol concentration. Because inhaled long-acting beta2 agonists and corticosteroid combination treatment produces better control of symptoms and lung function, with no greater risk of side-effects than that with use of either component alone, this combination treatment should be considered for patients with COPD.